Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Gut commensal Christensenella minuta modulates host metabolism via acylated secondary bile acids

Abstract

A strong correlation between gut microbes and host health has been observed in numerous gut metagenomic cohort studies. However, the underlying mechanisms governing host–microbe interactions in the gut remain largely unknown. Here we report that the gut commensal Christensenella minuta modulates host metabolism by generating a previously undescribed class of secondary bile acids with 3-O-acylation substitution that inhibit the intestinal farnesoid X receptor. Administration of C. minuta alleviated features of metabolic disease in high fat diet-induced obese mice associated with a significant increase in these acylated bile acids, which we refer to as 3-O-acyl-cholic acids. Specific knockout of intestinal farnesoid X receptor in mice counteracted the beneficial effects observed in their wild-type counterparts. Finally, we showed that 3-O-acyl-CAs were prevalent in healthy humans but significantly depleted in patients with type 2 diabetes. Our findings indicate a role for C. minuta and acylated bile acids in metabolic diseases.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Gut microbe-mediated biotransformation of CAs into previously unknown 3-O-acyl-CAs.
Fig. 2: The effects of C. minuta on the host metabolism in DIO mice.
Fig. 3: The effects of C. minuta on the intestinal FXR–enterohepatic signalling axis in DIO mice.
Fig. 4: The cell culture-based identification of the physiological function and the germ-free mouse-based simple pharmacokinetics of 3-O-acyl-CAs.
Fig. 5: The effects of C. minuta on HFD-induced intestinal FxrΔIE mice and their wild-type counterparts (Fxrfl/fl).
Fig. 6: The distribution and concentration of 4 newly identified 3-O-acyl-CAs and 12 common BAs in the faeces of the T2D cohort (n = 46 biologically independent individuals).

Similar content being viewed by others

Data availability

The datasets generated in this study were deposited in NCBI under Bioproject PRJNA834918. The accession number of the C. minuta genome is JAMBPM000000000. The accession number of the 16S rDNA amplicon data is SRR26844427. The output of the quantitative metabolomics is available in Supplementary Table 15. The seven publicly available datasets used for the identification of species depleted in metabolic disease cohorts are numbered as MD_1 to MD_7, with accession numbers of PRJEB4336 (phenotype: obesity and health), PRJEB12357 (phenotype: obesity and health), PRJEB12123 (phenotype: obesity and health), PRJEB21528 (phenotype: CVD and health), PRJNA422434 (phenotype: type 2 diabetes and health), PRJEB1786 (phenotype: type 2 diabetes and health) and PRJNA373901 (phenotype: NAFLD and health), respectively. Source data are provided with this paper.

References

  1. Koh, A. & Backhed, F. From association to causality: the role of the gut microbiota and its functional products on host metabolism. Mol. Cell 78, 584–596 (2020).

    Article  CAS  PubMed  Google Scholar 

  2. Fan, Y. & Pedersen, O. Gut microbiota in human metabolic health and disease. Nat. Rev. Microbiol. 19, 55–71 (2021).

    Article  CAS  PubMed  Google Scholar 

  3. Wahlstrom, A., Sayin, S. I., Marschall, H. U. & Backhed, F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 24, 41–50 (2016).

    Article  PubMed  Google Scholar 

  4. Fiorucci, S. & Distrutti, E. Bile acid-activated receptors, intestinal microbiota, and the treatment of metabolic disorders. Trends Mol. Med. 21, 702–714 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Winston, J. A. & Theriot, C. M. Diversification of host bile acids by members of the gut microbiota. Gut Microbes 11, 158–171 (2020).

    Article  PubMed  Google Scholar 

  6. Kuipers, F., Bloks, V. W. & Groen, A. K. Beyond intestinal soap-bile acids in metabolic control. Nat. Rev. Endocrinol. 10, 488–498 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. Islam, K. B. M. S. et al. Bile acid is a host factor that regulates the composition of the cecal microbiota in rats. Gastroenterology 141, 1773–1781 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Gérard, P. Metabolism of cholesterol and bile acids by the gut microbiota. Pathogens 3, 14–24, (2014).

  9. Quinn, R. A. et al. Global chemical effects of the microbiome include new bile-acid conjugations. Nature 579, 123–129 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Funabashi, M. et al. A metabolic pathway for bile acid dehydroxylation by the gut microbiome. Nature 582, 566–570 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lucas, L. N. et al. Dominant bacterial phyla from the human gut show widespread ability to transform and conjugate bile acids. mSystems 6, e00805–e00821, (2021).

  12. Sato, Y. et al. Novel bile acid biosynthetic pathways are enriched in the microbiome of centenarians. Nature 599, 458–464 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  13. Hang, S. Y. et al. Bile acid metabolites control Th17 and Treg cell differentiation. Nature 576, 143–148 (2020).

    Article  ADS  Google Scholar 

  14. Goodrich, J. K. et al. Human genetics shape the gut microbiome. Cell 159, 789–799 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Li, X. et al. Regional distribution of Christensenellaceae and its associations with metabolic syndrome based on a population-level analysis. PeerJ 8, e9591 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Waters, J. L. & Ley, R. E. The human gut bacteria Christensenellaceae are widespread, heritable, and associated with health. BMC Biol. 17, 83 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Mazier, W. et al. A new strain of christensenella minuta as a potential biotherapy for obesity and associated metabolic diseases. Cells 10, 823 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Pan, T. et al. Christensenella regulated by Huang-Qi-Ling-Hua-San is a key factor by which to improve type 2 diabetes. Front. Microbiol. 13, 1022403 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Liu, C. et al. Enlightening the taxonomy darkness of human gut microbiomes with a cultured biobank. Microbiome 9, 119 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Koh, A., De Vadder, F., Kovatcheva-Datchary, P. & Backhed, F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial Metabolites. Cell 165, 1332–1345 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Charrier, C. et al. A novel class of CoA-transferase involved in short-chain fatty acid metabolism in butyrate-producing human colonic bacteria. Microbiology 152, 179–185 (2006).

    Article  CAS  PubMed  Google Scholar 

  22. Yao, L. et al. High-level production of γ-aminobutyric acid via efficient co-expression of the key genes of glutamate decarboxylase system in Escherichia coli. Eng. Microbiol. 3, 100077 (2023).

    Article  CAS  Google Scholar 

  23. Yang, Y. Y., Gu, H. W., Sun, Q. L. & Wang, J. F. Effects of Christensenella minuta lipopolysaccharide on RAW 264.7 macrophages activation. Microb. Pathog. 125, 411–417 (2018).

    Article  CAS  PubMed  Google Scholar 

  24. Thomas, C. et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. 10, 167–177 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Duboc, H., Tache, Y. & Hofmann, A. F. The bile acid TGR5 membrane receptor: from basic research to clinical application. Dig. Liver Dis. 46, 302–312 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zheng, X. J. et al. Hyocholic acid species improve glucose homeostasis through a distinct TGR5 and FXR signaling mechanism. Cell Metab. 33, 791–803 (2021).

    Article  CAS  PubMed  Google Scholar 

  27. Kliewer, S. A. & Mangelsdorf, D. J. Bile acids as hormones: the FXR–FGF15/19 pathway. Dig. Dis. 33, 327–331 (2015).

    Article  PubMed  Google Scholar 

  28. Hla, T. & Kolesnick, R. C16:0–ceramide signals insulin resistance. Cell Metab. 20, 703–705 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Xie, C. et al. An intestinal farnesoid X receptor-ceramide signaling axis modulates hepatic gluconeogenesis in mice. Diabetes 66, 613–626 (2017).

    Article  CAS  PubMed  Google Scholar 

  30. Pagadala, M., Kasumov, T., McCullough, A. J., Zein, N. N. & Kirwan, J. P. Role of ceramides in nonalcoholic fatty liver disease. Trends Endocrinol. Metab. 23, 365–371 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Galadari, S. et al. Role of ceramide in diabetes mellitus: evidence and mechanisms. Lipids Health Dis. 12, 98 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Tavella, T. et al. Elevated gut microbiome abundance of Christensenellaceae, Porphyromonadaceae and Rikenellaceae is associated with reduced visceral adipose tissue and healthier metabolic profile in Italian elderly. Gut Microbes 13, 1–19 (2021).

    Article  PubMed  Google Scholar 

  33. Vital, M., Rud, T., Rath, S., Pieper, D. H. & Schlüter, D. Diversity of bacteria exhibiting bile acid-inducible 7α-dehydroxylation genes in the human gut. Comput. Struct. Biotechnol. J. 17, 1016–1019 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Miyamoto, J. et al. Gut microbiota confers host resistance to obesity by metabolizing dietary polyunsaturated fatty acids. Nat. Commun. 10, 4007 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  35. Zhang, Y. J. et al. Ursodeoxycholic acid alters bile acid and fatty acid profiles in a mouse model of diet-induced obesity. Front. Pharm. 10, 842 (2019).

    Article  CAS  Google Scholar 

  36. Wang, K. et al. Parabacteroides distasonis alleviates obesity and metabolic dysfunctions via production of succinate and secondary bile acids. Cell Rep. 26, 222–235 (2019).

    Article  PubMed  Google Scholar 

  37. Qiao, S. et al. Activation of a specific gut Bacteroides–folate–liver axis benefits for the alleviation of nonalcoholic hepatic steatosis. Cell Rep. 32, 108005 (2020).

    Article  CAS  PubMed  Google Scholar 

  38. Yoshida, N. et al. Bacteroides vulgatus and Bacteroides dorei reduce gut microbial lipopolysaccharide production and inhibit atherosclerosis. Circulation 138, 2486–2498 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Wu, C. et al. Strain-level screening of human gut microbes identifies blautia producta as a novel anti-hyperlipidemic probiotic via the production of 12-methylmyristic acid. Gut Microbes 15, 2228045 (2021).

    Google Scholar 

  40. Everard, A. et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl Acad. Sci. USA 110, 9066–9071 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  41. Plovier, H. et al. A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nat. Med. 23, 107–113 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Zhao, S. et al. Akkermansia muciniphila improves metabolic profiles by reducing inflammation in chow diet-fed mice. J. Mol. Endocrinol. 58, 1–14 (2017).

    Article  PubMed  Google Scholar 

  43. Sun, L. L. et al. Gut microbiota and intestinal FXR mediate the clinical benefits of metformin. Nat. Med. 24, 1919–1929 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wu, Q. et al. Suppressing the intestinal farnesoid X receptor/sphingomyelin phosphodiesterase 3 axis decreases atherosclerosis. J. Clin. Invest. 131, e142865 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Russell, B. J. et al. Intestinal transgene delivery with native E. coli chassis allows persistent physiological changes. Cell 185, 3263–3277 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Clifford, B. L. et al. FXR activation protects against NAFLD via bile-acid-dependent reductions in lipid absorption. Cell Metab. 33, 1671–1684 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Jiang, C. et al. Intestine-selective farnesoid X receptor inhibition improves obesity-related metabolic dysfunction. Nat. Commun. 6, 10166 (2015).

    Article  ADS  CAS  PubMed  Google Scholar 

  48. Yamagata, K. et al. Bile acids regulate gluconeogenic gene expression via small heterodimer partner-mediated repression of hepatocyte nuclear factor 4 and Foxo1. J. Biol. Chem. 279, 23158–23165 (2004).

    Article  CAS  PubMed  Google Scholar 

  49. Jiang, C. et al. Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J. Clin. Invest. 125, 386–402 (2015).

    Article  PubMed  Google Scholar 

  50. Fang, Y., Hegazy, L., Finck, B. N. & Elgendy, B. Recent advances in the medicinal chemistry of farnesoid X receptor. J. Med. Chem. 64, 17545–17571 (2021).

    Article  CAS  PubMed  Google Scholar 

  51. Fu, T. et al. FXR regulates intestinal cancer stem cell proliferation. Cell 176, 1098–1112 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Gadaleta, R. M. et al. Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease. Gut 60, 463–472 (2011).

    Article  CAS  PubMed  Google Scholar 

  53. Riley, D. J., Kyger, E. M., Spilburg, C. A. & Lange, L. G. Pancreatic cholesterol esterases. 2. Purification and characterization of human pancreatic fatty acid ethyl ester synthase. Biochemistry 29, 3848–3852 (1990).

    Article  CAS  PubMed  Google Scholar 

  54. Virtue, S. & Vidal-Puig, A. GTTs and ITTs in mice: simple tests, complex answers. Nat. Metab. 3, 883–886 (2021).

    Article  PubMed  Google Scholar 

  55. Fujii, H. et al. Expression of perilipin and adipophilin in nonalcoholic fatty liver disease; relevance to oxidative injury and hepatocyte ballooning. J. Atheroscler. Thromb. 16, 893–901 (2009).

    Article  CAS  PubMed  Google Scholar 

  56. Gomez, C., Stucheli, S., Kratschmar, D. V., Bouitbir, J. & Odermatt, A. Development and validation of a highly sensitive LC–MS/MS Method for the analysis of bile acids in serum, plasma, and liver tissue samples. Metabolites 10, 282 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Yang, T. et al. Quantitative profiling of 19 bile acids in rat plasma, liver, bile and different intestinal section contents to investigate bile acid homeostasis and the application of temporal variation of endogenous bile acids. J. Steroid Biochem. Mol. Biol. 172, 69–78 (2017).

    Article  CAS  PubMed  Google Scholar 

  58. Jiang, Z. L. et al. Dietary fruit and vegetable intake, gut microbiota, and type 2 diabetes: results from two large human cohort studies. Bmc Med 18, 371 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Li, D. H. et al. Alkaliphilus flagellatus sp. nov., Butyricicoccus intestinisimiae sp. nov., Clostridium mobile sp. nov., Clostridium simiarum sp. nov., Dysosmobacter acutus sp. nov., Paenibacillus brevis sp. nov., Peptoniphilus ovalis sp. nov. and Tissierella simiarum sp. nov., isolated from monkey faeces. Int J. Syst. Evol. Microbiol. 72, 005276 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Zhao, H. et al. High-yield oleaginous fungi and high-value microbial lipid resources from Mucoromycota. Bioenerg. Res. 14, 1196–1204 (2020).

    Article  Google Scholar 

  61. Chen, B. S. et al. Lanostane triterpenoids with glucose-uptake-stimulatory activity from peels of the cultivated edible mushroom Wolfiporia cocos. J. Agr. Food Chem. 67, 7348–7364 (2019).

    Article  ADS  CAS  Google Scholar 

  62. King, T. P. Separation of proteins by ammonium sulfate gradient solubilization. Biochemistry 11, 367–371 (1972).

    Article  CAS  PubMed  Google Scholar 

  63. Chen, S. F., Zhou, Y. Q., Chen, Y. R. & Gu, J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 34, 884–890 (2018).

    Article  Google Scholar 

  64. Segata, N. et al. Metagenomic biomarker discovery and explanation. Genome Biol. 12, R60 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Ultra-fast sequence analysis v.11 (USEARCH, 2013); https://drive5.com/usearch/

  66. Quast, C. et al. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 41, D590–D596 (2013).

    Article  CAS  PubMed  Google Scholar 

  67. MicrobiomeAnalyst v.2.0 (MicrobiomeAnalyst, 2022); https://www.microbiomeanalyst.ca/MicrobiomeAnalyst/

  68. Chong, J., Liu, P., Zhou, G. Y. & Xia, J. G. Using MicrobiomeAnalyst for comprehensive statistical, functional, and meta-analysis of microbiome data. Nat. Protoc. 15, 799–821 (2020).

    Article  CAS  PubMed  Google Scholar 

  69. Sun, T. et al. 3MCor: an integrative web server for metabolome-microbiome-metadata correlation analysis. Bioinformatics 38, 1378–1384 (2022).

    Article  CAS  PubMed  Google Scholar 

  70. Kohl, M., Wiese, S. & Warscheid, B. Cytoscape: software for visualization and analysis of biological networks. Methods Mol. Biol. 696, 291–303 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported financially by the National Key Research and Development Program of China and The National Science Fund for Distinguished Young Scholars (2021YFA0717002, 2022YFA1304103, 31925021, 2019YFA0905601 and 2018YFA0800701).

Author information

Authors and Affiliations

Authors

Contributions

C.L. performed the in silico analysis. M.-X.D., L.-S.X., Y.J., C.-Y.Y., M.-Z.J. and S.-S.Q. performed the animal trials and related tests. C.L., M.-X.D., L.-S.X., X.-W.S. and L.-S.W. performed the in vitro experiments. C.L., B.-S.C. and W.-Z.W. characterized the new bile acids. C.L., X.L., K.W., Y.J. and D.W. performed the cell experiments. M.S., B.-J.C., X.-W.S. and H.-J.H. performed the cohort experiments; W.-Z.W, S.-P.Q. and C.-K.L. performed the LC–MS analysis. S.-J.L. and C.L. designed the experiments. S.-J.L., H.-W.L. and C.J. supervised and directed this project. C.L., M.-X.D., L.-S.X. and S.-J.L. analysed the data and wrote the paper. All authors commented on the paper.

Corresponding authors

Correspondence to Changtao Jiang, Hong-Wei Liu or Shuang-Jiang Liu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Microbiology thanks Andrew Patterson, Amir Zarrinpar and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 The LC-MS/MS spectra of the acylated cholic acid derivates.

a, The m/z size of characteristic product ion and the collision-induced dissociation (CID) energy was illustrated in the panels. b, Extracted ion chromatogram of four new BA compounds (marked as 1, 2, 3, 4 in the panel) produced by C. minuta cells during in-vitro transformation (CM culture) and the retention time alignments with standards of 3’-Acetylcholic acid (Ac-CA, m/z -H: 449.2909), 3’-Propionylcholic acid (Prp-CA, m/z -H: 463.3065), 3’-Butyrylcholic acid (Buty-CA, m/z -H: 477.3222) and 3’-Valerylcholic acid (Val-CA, m/z -H: 491.3378), which were determined by nuclear magnetic resonance (See Supplementary Information Data for more information about NMR analysis). c, Comparison of high-resolution primary mass spectra of four new BAs in C. minuta culture with standards. The formula, calculated mass, measured accurate mass of the standard and culture sample, the mass error in ppm and MS/MS fragmentation similarity score for each new BA were listed in the Supplementary information Table 2.

Extended Data Fig. 2 The LC-MS spectra displaying that the consumption of CA and the production of 4 unknown CA derivates by in vitro C. minuta biotransformation assay.

a and b, The TIC scan and EIC scan results for the broth of C. minuta transformation at 0 hour (a) and 24 hour (b) in the absence of SCFA mixture in the MMGMB media. c and d, The TIC scan and EIC scan results for the broth of C. minuta transformation at 0 hour (c) and 24 hour (d) in the presence of 1 mM SCFA mixture in the MMGMB media. The peak aera for CA (EIC = 407.3 m/z) and four known derivates (EIC = 449.3, 463.3, 477.3 and 491.3, respectively) were marked in the panels, and was used for calculation of CA transformation rate.

Extended Data Fig. 3 The in-vitro biotransformation activity of C. minuta.

a, The production of SCFAs during in vitro cultivation; STD SCFA: standard of SCFAs mixture (1 mM); CM culture: C. minuta cultivated in mGAM liquid media for 24 hours. The peak aera of each kind of SCFA were listed in the Supplementary Information Table 3. b, The diagram displaying the generation of 3-O-acyl-CAs catalyzed by C. minuta cells, cell lysates and crude proteins with different substrates.

Extended Data Fig. 4 The effects of C. minuta on DIO mice.

a, The ileal leptin; b, Plasma low density lipoprotein cholesterol (LDL); c, Liver’s index; d, Plasma aspartate amino transferase (AST); e, Plasma alanine aminotransferase (ALT); f, Hepatic aspartate amino transferase (AST); g, Hepatic alanine aminotransferase (ALT); h, The Plasma lipopolysaccharide (LPS); i, Ileum glucagon-like peptide 1 (GLP-1); j, Colonic GLP-1; k, The quantification of bile acids in ileal contents; l, The transcription level of bile acid receptor genes in ileum; m, The transcription level of bile acid receptor genes in colon; n, The transcription level of genes involved in hepatic circadian clock; o, The transcription level of bile acid related genes in liver. For panel k - o, the group names were marked with different color bars in the panel. DIO_CK group: DIO mice were fed with HFD and daily administrated with PBS buffer, n = 5 biologically independent samples; DIO_ND group: DIO mice (purchased from vendor) were fed with low fat normal control diet and daily administrated with PBS buffer for 8 weeks, n = 5 biologically independent samples; C57BL/6J group: normal C57BL/6J mice were fed with normal chow diet and daily administrated with PBS buffer, n = 5 biologically independent samples; DIO_CM group: DIO mice were fed with HFD and daily administrated with C. minuta cells (5*108 CFU), n = 10 biologically independent samples. For panel a, c, d, e, h, i and j, one-Way ANOVA test followed by Dunnett’s multiple comparisons test was performed for statistical analysis of difference between DIO_CK (the control group) and the other groups; For panel b, f, g, l, m, n and o, Kruskal-Wallis test followed by Dunn’s multiple comparisons test was performed for statistical analysis of difference between DIO_CK (the control group) and the other groups. Symbols: **** indicates P value < 0.0001, *** indicates P value < 0.001, ** indicates P value < 0.01, * indicates P value < 0.05. Data were shown as the mean ± SEM unless otherwise indicated.

Source data

Extended Data Fig. 5 The effects of C. minuta on host metabolism and metabolically-important hormones in DIO mice.

a, The averaged food consumption of each mouse per day; b, The changes of body weight during 7-week treatment; c, Free-diet blood glucose; d, Fasted-diet blood glucose; e and f, Blood glucose profile(e) and the AOC (f) calculated during the oral glucose tolerance test (OGTT); g and h, Blood glucose profile (g) and the aera of the curve (AOC) (h) calculated during the insulin tolerance test (ITT); i, Plasma triglycerides (TG) j, Plasma total cholesterol (TCHO); k, Active GLP-1 stimulus-secretion test; l, Serum insulin secretion test; m, Plasma glucagon; n, Glucose dependent insulin stimulating peptide (GIP); o, Plasma leptin; p, Plasma ghrelin. The physiological and biochemical indexes in panel c-l were measured using venous blood collected before mouse execution while the others were measured after the end-point execution and sampling of mice after 7-week treatments. DIO_CKrepeat group: DIO mice fed with HFD and daily administrated with PBS buffer for 7 weeks, n = 6 biologically independent samples; DIO_CMrepeat group: DIO mice fed with HFD and daily administrated with C. minuta cells (2*109 CFU) for 7 weeks, n = 6 biologically independent samples. C57BL/6Jrepeat group: normal C57BL/6J mice fed with normal control diet and daily administrated with PBS buffer for 7 weeks, n = 5 biologically independent samples; For panels a, Welch’s ANOVA test followed by two-sided Dunnett’s T3 multiple comparisons test was performed for statistical analysis of difference between DIO_CKrepeat (the control group) and the other groups; For panel b, d, f, g, h, i, j, l, and m, one-Way ANOVA test followed by Dunnett’s multiple comparisons test was performed for statistical analysis of difference between DIO_CKrepeat (the control group) and the other groups; For panel c, e, k, n, o and p, Kruskal-Wallis test followed by Dunn’s multiple comparisons test was performed for statistical analysis of difference between DIO_CKrepeat (the control group) and the other groups. Data were shown as the mean ± SEM unless otherwise indicated.

Source data

Extended Data Fig. 6 The microbiota and metabolome of caecal contents of experimental mice.

a and b, the box-and-whiskers plots displaying alpha diversity with Shannon index (a) and Chao1 index (b); statistical method: two-sided T test/ANOVA; c and d, the pCoA plots displaying the beta diversity; distance method: Bray-Curtis Index, statistical method: ANOSIM; e and f, The histograms displaying the LDA Effect Size (LefSe) analysis results of significantly differed taxonomic compositions at known taxonomic levels (e) and amplicon sequence variant (ASV) level (f); g, The volcano plot displaying the differed metabolite compositions of intestinal contents between DIO_CM and DIO_CK mice; Two-tail T test was used for statistical analysis of the significance of fold changes (FCs). The solid circles were colored by their metabolite categories as listed in the top of the panel; the 25 significantly-changed metabolites between DIO_CM and DIO_CK groups (P value < 0.05) were listed in the panel; h, The bar chart displaying the total concentrations of each chemical class that were significantly changed in panel g; i, The bar chart displaying the concentration of TUDCA, GUDCA and T-βMCA in the caecal contents; j, The correlation network displaying the spearman correlations between the changes of ASVs and gut metabolites; The square nodes represented the significantly changed metabolites in panel g and colored in terms of their chemical class, while the circle nodes represented the significantly changed ASVs listed in panel e; enriched metabolites/ASVs: pink color, reduced metabolites/ASVs: green color; The edge thickness manifested the correlation r values (the minimal cut-off value was 0.7), and the colors displayed the positive (pink) and negative (green) correlations between ASVs and metabolites. n = 5 biologically independent samples for DIO_CK, DIO_ND and C57BL/6J groups; n = 10 biologically independent samples for DIO_CM group. For panel h and i, two-tail T test was performed for statistical analysis of difference between DIO_CK and DIO_CM group; Symbols: **** indicates P value < 0.0001, *** indicates P value < 0.001, ** indicates P value < 0.01, * indicates P value < 0.05. Data were shown as the mean ± SEM unless otherwise indicated.

Source data

Extended Data Fig. 7 The multiple reaction monitoring (MRM) chromatograms of 3’-Acetylcholic acid (Ac-CA, a), 3’-Propionylcholic acid (Prp-CA, b), 3’-Butyrylcholic acid (Buty-CA, c) and 3’-Valerylcholic acid (Val-CA, d) and the retention time alignments of the standard (5 mg/ml), a representative mouse ileal content sample and a representative human fecal sample.

The high-resolution primary mass spectrum of each parent ion was provided at the top left corner in each panel. The accurate mass and mass error (ppm) of four compounds in the standard and each measured sample were listed in the Supplementary information Table 6. See Supplementary information Table 7 and Supplementary Information Table 13 for more detailed parameters and conditions used in the MRM methods.

Extended Data Fig. 8 The effects of 3-O-acyl-CAs on cell cultures.

a, The Dual-luciferase assay exhibiting the activation effect of FXR by 3-O-acyl-CAs, n = 4 biologically independent samples; b, The evaluation of cytotoxin of Ac-CA to Caco-2 cells with cell counting kit-8, n = 3 biologically independent samples; c, The evaluation of cytotoxin of Buty-CA to HEK293 cells with cell counting kit-8, n = 3 biologically independent samples; d, The evaluation of cytotoxin of Ac-CA to Caco-2 cells with cell counting kit-8, n = 3 biologically independent samples; e, The evaluation of cytotoxin of Buty-CA to Caco-2 cells with cell counting kit-8, n = 3 biologically independent samples; f, the dose–response inhibition of FXR by TβMCA, n = 3 biologically independent samples; g, the half maximal inhibitory concentrations (IC50) of TβMCA to the FXR; h, The Dual-luciferase assay exhibiting the effect of TGR5, n = 4 biologically independent samples; i and j, the distribution of CAs in germfree mice gavaged with 10 mg/kg body weight buty-CAs (g, n = 3 biologically independent samples) and in conventional C57BL/6J (h, n = 5 biologically independent samples). For panel a to f and h, one-Way ANOVA test followed by Dunnett’s multiple comparisons test was performed for statistical analysis of difference between control group (or 0 time point group) and the other groups; For panel i, Kruskal-Wallis test followed by Dunn’s multiple comparisons test was performed for statistical analysis of difference between the control group or 0 h group and the other groups. Symbols: **** indicates P value < 0.0001, *** indicates P value < 0.001, ** indicates P value < 0.01, * indicates P value < 0.05. Not significant groups were not marked in the panels. Data were shown as the mean ± SEM unless otherwise indicated.

Source data

Extended Data Fig. 9 The distribution of bile acids in germ-free mice gavaged with 10 mg/kg body weight buty-CAs after different time period.

The time point that the mice were sacrificed for analysis was marked in the panels with different colors of bars; n = 3 biologically independent samples; Kruskal-Wallis test followed by Dunn’s multiple comparisons test was performed for statistical analysis of difference between the control group or 0 h group and the other groups. Not significant groups were not marked in the panels. Data were shown as the mean ± SEM unless otherwise indicated.

Source data

Extended Data Table 1 The representative gut microbes for screening of BA transformation activity

Supplementary information

Supplementary Information

Supplemenetary Figs. 1 and 2, data and Tables 1–15. Supplementary Fig. 1. The taxonomic diversity of the 175 species that were negatively associated with metabolic diseases. a, The phylogenetic tree of the 175 gut microbial species that were depleted in at least one of the investigated seven cohort studies with metabolic diseases and their distributions in all the assessed gut metagenomic samples. The phylogenetic tree was constructed using the 16S rRNA gene sequences by the neighbour-joining method with 1,000 bootstraps. b, A heatmap displaying the LDA score (log10) of 175 species enriched in the healthy cohort compared with diseased counterparts for each cohort study (MD_1 to MD_7) based on LEfSe analysis. The 87 species for which we did not have cultivable representatives are marked as ‘NA’ in front of their Latin name. The colour depth of the heatmap indicates the LDA score (log10) of each species for study MD_1 to MD_7. The accession number of each study is available in Data Availability. Supplementary Fig. 2. LC–MS spectra showing that the C. minuta catalyse diverse BA compounds into corresponding acylation conjugates. a–g, C. minuta-mediated acylation of CA (a), CCA (b), TCA (c), CDCA (d), GCDCA (e), TCDCA (f) and DCA (g). The extracted-ion chromatogram scan-based peak area and m/z size of the BAs and their acylated derivates was marked in the panels. Supplementary data. The results of nuclear magnetic resonance-based characterization of 3-O-acyl-CAs.

Reporting Summary

41564_2023_1570_MOESM3_ESM.xlsx

Supplementary Table 1. The representative gut microbes for screening of BA transformation activity. Supplementary Table 2. The accurate mass and errors of the standards and metabolites in samples. Supplementary Table 3. The production of SCFAs during in vitro cultivation. Supplementary Table 4. The information of proteins in the purified catalytic protein components. Supplementary Table 5. The abundance and taxonomy table of ASVs. The reads abundance for each sample were nomolized to the minimum size of 106,179. Supplementary Table 6. The accurate mass and mass error (ppm) of four compounds in the standard and each measured sample. Supplementary Table 7. The MRM-based quantification of bile acids in vivo. Supplementary Table 8. The basic information of faecal donors in the cohort. Supplementary Table 9. The commercial kits used in this study. Supplementary Table 10. Primers and annealing temperatures of the real time qPCR assays. Supplementary Table 11. Basic information of bile acids detected by LC–MS. Supplementary Table 12. The gradient elution pattern used for LC–MS analysis in this study. Supplementary Table 13. MRM method characteristics and validation parameters for the quantification of bile acids in liver tissue by LC–MS/MS. Supplementary Table 14. The accessions and group of each valid runs used for in silico analysis. Supplementary Table 15. The output of quantitative metabolomics.

Source data

Source Data Fig. 1

LC–MS/MS spectra without data for Fig. 1d.

Source Data Fig. 2

Source Data Fig. 2.

Source Data Fig. 3

Schematic diagram without data for Fig. 3a.

Source Data Fig. 4

Source Data Fig. 4

Source Data Fig. 5

Source Data Fig. 5

Source Data Fig. 6

Source Data Fig. 6

Source Data Extended Data Fig. 4

Source Data Extended Data Fig. 4

Source Data Extended Data Fig. 5

Source Data Extended Data Fig. 5

Source Data Extended Data Fig. 6

Extended Data Fig. 6a–f are the outputs of in silico analysis of 16S rDNA amplicon data. The raw ASV table is given in Supplementary Table 5. Extended Data Fig. 6g–i are the result of metabolomics. The raw data table is provided as Supplementary Table 15

Source Data Extended Data Fig. 8

Source Data Extended Data Fig. 8

Source Data Extended Data Fig. 9

Source Data Extended Data Fig. 9

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, C., Du, MX., Xie, LS. et al. Gut commensal Christensenella minuta modulates host metabolism via acylated secondary bile acids. Nat Microbiol 9, 434–450 (2024). https://doi.org/10.1038/s41564-023-01570-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-023-01570-0

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing